CN113813364A - Anti-addiction and anti-relapse application of polypeptide, complex and polypeptide - Google Patents

Anti-addiction and anti-relapse application of polypeptide, complex and polypeptide Download PDF

Info

Publication number
CN113813364A
CN113813364A CN202111321033.2A CN202111321033A CN113813364A CN 113813364 A CN113813364 A CN 113813364A CN 202111321033 A CN202111321033 A CN 202111321033A CN 113813364 A CN113813364 A CN 113813364A
Authority
CN
China
Prior art keywords
polypeptide
seq
relapse
addiction
substance addiction
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202111321033.2A
Other languages
Chinese (zh)
Inventor
谭震
李涛
李书鹏
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shenzhen Chenyang Biotechnology Co ltd
Original Assignee
Shenzhen Chenyang Biotechnology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shenzhen Chenyang Biotechnology Co ltd filed Critical Shenzhen Chenyang Biotechnology Co ltd
Priority to CN202111321033.2A priority Critical patent/CN113813364A/en
Publication of CN113813364A publication Critical patent/CN113813364A/en
Priority to PCT/CN2022/117272 priority patent/WO2023082805A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70507CD2

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Addiction (AREA)
  • Organic Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Molecular Biology (AREA)
  • Psychiatry (AREA)
  • Biomedical Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Cell Biology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The invention relates to the anti-addiction and relapse use of polypeptide, and provides the use of polypeptide in preparing medicine for treating and/or preventing substance addiction or relapse of substance addiction, wherein the polypeptide consists of at least 11 continuous amino acid residues in a sequence shown as SEQ ID NO. 1 and contains a sequence shown as SEQ ID NO. 2. The polypeptide of the invention has better clinical development value. The invention also relates to the use of the polypeptides, nucleic acid molecules, expression vectors, host cells and complexes containing the polypeptides.

Description

Anti-addiction and anti-relapse application of polypeptide, complex and polypeptide
Technical Field
The present invention relates to the field of substance addiction, in particular to the use of polypeptides in the prevention and treatment of addiction or relapse of addiction.
Background
The World Health Organization (WHO) is defined as substance addiction: despite the understanding and experience of the deleterious effects of the substance, the substance is used repeatedly. Substance addiction is a chronic, recurrent disease characterized by uncontrolled use of a substance, compulsive foraging and craving of a substance, consistent use regardless of adverse consequences, and physical and/or psychological dependence on a substance. In general, substance addiction follows the course of tolerance, withdrawal, compulsive medication, foraging, implementation of addictive behaviors, and relapse.
Substance addiction has become a global medical and social concern that has significant social and economic impact on both addicts and society. For example, substance addiction is often linked to the spread of violent crimes and infectious diseases. For another example, substance addiction can result in partial or complete loss of the addict's workforce, thereby negatively impacting individuals, families, and society. In particular, as one of the most common abused substances worldwide, alcoholism causes severe liver and cardiovascular diseases and is prone to severe mental disorders, social problems and adverse consequences, including family breakdowns, tragic accidents and reduced work efficiency.
In the case of drug rehabilitation or withdrawal treatment of addicts, some withdrawal drugs are often used, but these drugs still have limitations.
For example, in the case of addiction to opioids, the following approaches are often used: (i) replacement therapy, replacement with methadone is currently the most common method of detoxification; (ii) (ii) an opioid receptor antagonist, naltrexone or naloxone, to promote withdrawal; (c) non-opioid treatment with clonidine and lofexidine to suppress withdrawal symptoms. While these approaches may render the addict no longer exhibit physiological discomfort (e.g., chills, vomiting, tremor, etc.) upon withdrawal of the addict, studies have shown that these drugs do not reduce the mental dependence (or psychological dependence) of the addict.
As another example, different classes of drugs (e.g., naltrexone, acamprosate, ondansetron, disulfiram, Y-hydroxybutyric acid (GHB), and topiramate) have been tested for their potential therapeutic effects on alcohol addiction. Several of these pharmacotherapeutic agents such as naltrexone, acamprosate and disulfiram have demonstrated some efficacy and are approved for the treatment of alcoholism. Of these drugs, naltrexone is currently considered the pharmacologically preferred choice. However, despite some promising results, none of these drugs (including naltrexone) is sufficiently effective in alcoholism and the prognosis is still poor.
Accordingly, in the field of resistance to substance addiction, there is a strong need to develop safe and effective drugs to provide richer, better therapeutic or prophylactic strategies for substance addiction and relapse thereof.
Disclosure of Invention
In order to achieve the above purpose, the inventors have conducted extensive studies on the molecular mechanism of addiction, and found that a novel polypeptide is capable of disrupting the interaction between the D2 receptor and the NMDA receptor.
In particular, the DA receptor is a receptor located in the organism that acts through its corresponding membrane receptor. DA receptors can be divided into five types: d1, D2, D3, D4 and D5. The D2 receptor (D2R) is widely expressed in the brain.
NMDA receptors (NMDAR) not only play important physiological roles in the development of the nervous system, such as regulating neuronal survival, regulating neuronal dendrites, axon structure development, and participating in synaptic plasticity; and plays a key role in the formation of a neuron circuit and the pathogenesis of various neuropsychiatric diseases, and data show that the NMDA receptor is a key receptor in the learning and memory process. NMDAR is composed of mainly 3 different subunits, NR1, NR2 and NR3, respectively. Among them, the NR2 subunit has 4 different subunits, namely NR2A, NR2B, NR2C and NR 2D.
The inventors confirmed that the polypeptide of the present invention is effective for treating addiction to an addictive substance and inhibiting or treating relapse, thereby completing the present invention.
Accordingly, in a first aspect, the present invention provides a polypeptide consisting of at least 11 contiguous amino acid residues of the sequence shown as SEQ ID NO. 1 and comprising the sequence shown as SEQ ID NO. 2. In addition, the invention also provides the application of the polypeptide in preparing medicines for preventing and/or treating substance addiction.
As used herein, the term "treating" refers to causing a desired or beneficial effect in a patient, which may include reducing the frequency or severity of one or more symptoms of a disease, or suppressing or inhibiting the further development of a disease, disorder or condition.
As used herein, the term "preventing" refers to preventing or delaying the onset of a disease, or preventing the appearance of clinical or subclinical symptoms thereof.
As used herein, "substance addiction" refers to a chronic, recurrent disease characterized by uncontrolled use of a substance, compulsive foraging and craving of a substance, consistent use regardless of adverse consequences, and physical and/or psychological dependence on a substance. Substance addiction may, for example, be the first addiction to the substance, or refer to the formation and/or expression of substance addiction.
In exemplary embodiments, the polypeptide of the invention consists of at least 11, at least 12, at least 13, at least 14 or at least 15 consecutive amino acid residues of the sequence shown as SEQ ID No. 1.
In an exemplary embodiment, the sequence of the polypeptide of the invention consists of positions 1-15, 1-14, 1-13, 2-15, 2-14, 2-13, 3-15, 3-14 or 3-13 of SEQ ID NO 1 (KIYIVLRRRRKRVNT).
In a specific embodiment, the polypeptide of the invention has a sequence as shown in SEQ ID NO 1, SEQ ID NO 3 or SEQ ID NO 4.
In some embodiments, the addictive substance may be selected from the group consisting of: morphine, nicotine, alcohol, ***e, codeine, dihydrocodeine, hydromorphone, oxycodone, methadone, morphine, fentanyl, and meperidine.
In a second aspect, the invention provides the use of a polypeptide of the invention in the manufacture of a medicament for the prevention and/or treatment of relapse of substance addiction.
As used herein, "relapse," also referred to as "relapse," where appropriate, refers to the addictive substance used by the addict after withdrawal, but prior to the withdrawal. Relapse may be caused by environmental (situational), addictive substances, and/or stress (stress) factors.
In exemplary embodiments, recurrence in the present invention is due to environmental factors.
In an exemplary embodiment, the relapse in the present invention is caused by an addictive substance factor.
In an exemplary embodiment, the recurrence in the present invention is caused by a stressor.
The polypeptide of the invention is unexpectedly found to be capable of blocking the combination of the D2R/NR2B complex and eliminating the formation of addictive substances.
Moreover, the polypeptide of the present invention can eliminate relapse of addiction caused by environment, addictive substances, stress factors, and even produce statistically significant effects under the condition of high/low dose of addictive substance induction, and has good clinical development value.
In a third aspect, the invention provides a nucleic acid molecule encoding a polypeptide of the invention. Furthermore, the use of a nucleic acid molecule of the invention for the preparation of a medicament for the treatment and/or prevention of substance addiction or for the preparation of a medicament for the treatment and/or prevention of relapse of substance addiction is provided.
The nucleic acid molecules of the invention are used for producing the polypeptides of the invention, and the sequence of the nucleic acid molecules can be adjusted as appropriate by the person skilled in the art, depending on the expression system used.
In an exemplary embodiment, the nucleic acid molecule has the sequence shown as SEQ ID NO 5.
In a fourth aspect, the present invention provides an expression vector comprising a nucleic acid molecule of the invention. In addition, the invention also provides the application of the expression vector in preparing a medicament for treating and/or preventing substance addiction, or the application of the expression vector in preparing a medicament for treating and/or preventing substance addiction relapse.
The nucleic acid sequences of the invention may be inserted into expression vectors using a variety of known methods. For example, the nucleic acid molecule can be inserted into an appropriate restriction endonuclease site. Standard techniques for cloning, isolation, amplification and purification, enzymatic reactions involving DNA ligases, DNA polymerases, restriction endonucleases and the like in the procedures, and various isolation techniques are well known and commonly used by those skilled in the art.
In a fifth aspect, the invention provides a host cell comprising a nucleic acid molecule or expression vector of the invention. Furthermore, the use of a host cell of the invention for the preparation of a medicament for the treatment and/or prevention of substance addiction or the use of a host cell of the invention for the preparation of a medicament for the treatment and/or prevention of relapse of substance addiction is provided.
The polypeptides of the invention can be produced using a variety of expression systems, such as expression vectors and host cells in prokaryotic and eukaryotic expression systems. As will be described below with reference to mammalian expression systems, host cells may include the COS-7 cell line of monkey kidney fibroblasts and other cell lines capable of expressing compatible vectors, such as the C127, 3T3, CHO, HeLa and BHK cell lines. Mammalian expression vectors should contain an origin of replication, a suitable promoter and enhancer, and any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5' flanking nontranscribed sequences. DNA sequences derived from, for example, SV40 splicing and polyadenylation sites can be used to provide the required non-transcribed genetic elements. The expression vector may be introduced into the host cell by a variety of methods familiar to those skilled in the art, including, but not limited to, for example, calcium phosphate transfection, DEAE-dextran mediated transfection, or electroporation.
In a sixth aspect, the invention provides a complex comprising a polypeptide of the invention and a (transport) carrier attached thereto for permeabilizing the blood-brain barrier.
In exemplary embodiments, the carrier for permeabilizing the blood-brain barrier may be: HIV-1Tat protein, insulin, cationized albumin, monoclonal antibody (OX26) against mouse transferrin receptor, murine monoclonal antibody (HIRMAb) against human insulin receptor, Pennetratin, transduction domain of Tat protein, Pep-1 peptide, S413-one or more of PV, Magainin 2 and Buforin 2. For example, the TAT transduction domain can transduce intracellularly through a membrane and has the amino acid YGRKKRRQRRR (shown in SEQ ID NO: 6).
The polypeptides of the invention may be attached to a carrier for permeabilizing the blood-brain barrier by a suitable attachment technique. Exemplary linking techniques may be avidin-biotin techniques, polyethylene glycol (PEG) based space arm techniques, fusion protein techniques, and the like. For example, in the case of using the transduction domain of the HIV-1Tat protein as a carrier for permeability of the blood-brain barrier, the polypeptide of the invention may be directly linked to the transduction domain of the Tat protein by fusion protein technology.
In some embodiments, where fusion protein technology is used, a Linker (Linker) may also be used to attach the polypeptide of the invention to a carrier that is used to permeabilize the blood-brain barrier. Exemplary linkers may be glycine-bearing flexible linkers such as G, GSG, GSGGSG, GSGGSGG, GSGGSGGG, GGGGSGGG, ggggggs, SGG, and the like.
Drawings
FIG. 1 shows the results of co-immunoprecipitation of mouse hippocampal tissues;
fig. 2 shows the position of the fragment referred to in example 3 on D2R;
FIG. 3 shows the results of the GST-pulldown experiments for the fragments of example 3;
FIG. 4 shows co-immunoprecipitation results of KT polypeptide-treated CPP mice and BS mouse hippocampal tissues;
FIG. 5 shows the experimental protocol design of example 5;
FIG. 6 shows CPP scoring results of example 5;
FIG. 7 shows the experimental protocol design of example 6;
FIG. 8 shows the CPP scoring results of example 6; a: day 10, measurement results of each group; b: comparison of day 11 with day 10 of TAT-D2R-KT group;
FIG. 9 shows the experimental protocol design of example 7;
FIG. 10 shows the CPP measurement results on day 10 of example 7;
FIG. 11 shows the CPP measurement of test 1 of example 7;
FIG. 12 shows the CPP measurement results of test 2 of example 7;
FIG. 13 shows the CPP measurement of test 3 of example 7;
FIG. 14 shows the CPP measurement of test 4 of example 7;
FIG. 15 shows the experimental protocol design of example 8;
fig. 16 shows BS measurement results of test 1 of example 8; a: test 1 BS movement distance result; b: the BS movement distance result of test 2; c: test 3 BS movement distance result;
figure 17 shows the effect of different polypeptides prepared in example 9 on morphine-induced mouse CPP, where n-12; # #: p <0.01, relative to saline group; **: p <0.01, relative to the morphine group;
figure 18 shows the negativity of the different polypeptides prepared in example 9 to sensitization of mouse behavior by ***e, where n-10; *: p <0.05, x: p <0.01, relative to veh group;
figure 19 shows the negativity of the different polypeptides prepared in example 9 to sensitization of alcohol-induced mouse behaviour, where n-10; # #: p <0.01, relative to saline group; **: p <0.01, relative to the alcohol group.
Detailed Description
Currently, how to eliminate substance addiction or prevent relapse, thereby avoiding a significant burden on individuals, families, and society, has become a medical and social concern worldwide. Of these, it is interesting to note that Liu et al (Modulation of D2R-NR2B Interactions in Response to Cocaine, Neuron 52, 897-909, December 7,2006) reported that dopamine stimulation by Cocaine enhanced the formation of the heteromeric complex between the D2R and NMDA receptor NR2B subunits in new striatum in vivo; by studying the interaction of D2R with NR2B, the region of D2R responsible for the formation of the D2R-NR2B complex was found to be located at IL 3; further competition experiments showed that the motif containing ten residues in IL3 (TKRSSRAFRA, T225-a234) is critical for blocking the binding of D2R to NR 2B.
Notably, Liu et al tested a variety of polypeptides from IL3 of D2R and the results indicated (see figures 3M and 3N in this document): the polypeptides (P1, P2, P3 and P5) successfully blocked D2R from binding to NR2B only in the presence of the motif T225-a234 described above; in the absence of this motif, the polypeptide (P4, I210-K226) has no hindering effect; meanwhile, the control polypeptide P6 obtained by scrambling the sequence of the polypeptide P5 containing this motif similarly does not prevent the binding of D2R to NR 2B. Accordingly, Liu et al consider binding to be sequence specific and TKRSSRAFRA to be a potential binding motif.
However, the inventors have unexpectedly found in their studies that: in contrast to the report by Liu et al, the polypeptide K211-T225 in the IL3 region of D2R (which lacks one residue at each of the N-and C-termini; while not including the key motif TKRSSRAFRA, compared to P4, which was identified as being unable to block D2R from binding to the NR2B polypeptide) successfully reduced the interaction of D2R with NR 2B.
On the basis of further research work, the inventors confirmed that the polypeptide K211-T225 and N-terminal and C-terminal deletion variants thereof in the IL3 region of D2R are capable of reversing addictive behavior changes in an addiction model.
Throughout the specification, unless otherwise specifically noted, terms used herein should be understood as having meanings as commonly used in the art. Accordingly, unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. If there is a conflict, the present specification will control.
The advantages and various effects of the present invention will be more clearly apparent from the following detailed description of the embodiments of the present invention taken in conjunction with the examples. It will be understood by those skilled in the art that these specific embodiments and examples are for the purpose of illustrating the invention and are not to be construed as limiting the invention.
The examples, in which specific conditions are not specified, were conducted under conventional conditions or conditions recommended by the manufacturer. The reagents or apparatus used are not indicated to the manufacturer, but are conventional products which are commercially available.
Unless otherwise indicated, the experimental animals used in the examples were adult C57/BL6J male mice (8 weeks old, 25-30g in weight) purchased from the Guangzhou center for medical laboratory animals in China. Mice were housed under 12-hour light/12-hour dark cycles with free access to diet and water throughout the procedure. All mice were acclimated in the animal room for one week prior to experimental procedures.
The bands and morphological data were analyzed using ImageJ and image lab software (ImageJ1.30) and statistically analyzed using GraphPadPrism8 software in the examples. The sample size was selected as described previously (Arifin and Zahirudin, 2017; Mead et al, 2012). All experiments were independently repeated three times. No randomization/blind method was employed. Data are presented as mean ± SEM. Different groups used either one-way or two-way analysis of variance followed by posthoc Tukey multiple comparison tests. P <0.05 was considered significant. Unless otherwise stated, a: p <0.05, x: p <0.001, x: p <0.0001, and: p < 0.000001.
EXAMPLE 1 confirmation of in vivo formation of D2R/NR2B Complex
Co-immunoprecipitation operation:
co-immunoprecipitation was performed using protein samples from hippocampal tissue (100-500. mu.g of protein). NR2B and D1R or D2R were precipitated using anti-NR 2B and D2R, D2R or NR2B and 25. mu.l protein A/G + agarose bead slurry (Santa Cruz Biotechnology, sc-2001), respectively.
The proteins were then separated on an 8% SDS-PAGE gel and detected with mouse antibodies against NR2B and D2R. Protein was detected using HRP conjugated secondary antibodies and enhanced chemiluminescence. The strips were density analyzed using image lab software.
To assess whether D2R and NR2B could form a protein complex, the co-immunoprecipitation method described above was first followed and the possible interaction of D2R and NR2B in normal mouse hippocampal tissues was examined, and the results are shown in FIG. 1.
As can be seen from fig. 1, the D2R antibody successfully precipitated NR2B from the hippocampal protein mixture, whereas the D1R antibody did not precipitate NR2B, indicating a D2R/NR2B protein coupling.
Example 2 verification of the pathological significance of the Complex
Next, in order to determine whether the interaction state of D2R and NR2B is related to substance addiction, co-immunoprecipitation experiments were performed using morphine-induced positional preference (CPP) and Behavior Sensitization (BS) mouse hippocampal tissues as samples according to the method in example 1, and the results are shown in fig. 1.
As can be seen from FIG. 1, both D2R and NR2B conjugation were significantly increased in mouse hippocampal tissues of CPP and BS, confirming that the formation of D2R/NR2B complex may play a causative role in substance addiction.
EXAMPLE 3 search for binding sites for Complex to each other
In order to find a physical interaction site between D2R and NR2B, first, cDNA fragments of the CT region (T428-C443) of D2R and the third intracellular loop (IL3) region (K211-Q373) of D2R were obtained by a full-length cDNA clone of D2R (Genbank accession No.: M29066.1). These fragments were subcloned into the BamH1/EcoR1 or BamH1/Xho1 sites of pGEX-4T-3 plasmid (Youbao, cat # VT 1255). An initiating methionine residue and a stop codon were added as appropriate. All constructs were re-sequenced to confirm that splicing fusions were properly achieved. Coli BL21 live cells (Kangsheng Life technologies, no: KTSM104L) were used for expression, and GST fusion proteins containing the IL3 domain of D2R (GST-D2R-IL3) and the CT domain of D2R (GST-D2R-CT) were purified from the bacterial lysate. Wherein, the specific positions of the coding sequences of the IL3 region and the CT region on the D2R are shown in figure 2.
Mu.g of solubilized hippocampal extract was diluted with 1 XPBS/0.1% Triton X-100 and incubated with 20. mu.l of protein GST resin (transgenic Biotechnology, cat # N21220, Beijing, China) saturated with GST protein alone or 15. mu.g of prepared GST fusion protein overnight at 4 ℃. Beads were washed 1-8 times with 1 XPBS/0.1% Triton X-100. The bound proteins were eluted with 2 Xloading buffer, separated by SDS-PAGE, and immunoblotted with the respective antibodies, as shown in A in FIG. 3.
As can be seen from a in fig. 3, the IL3 region of D2R affinity precipitated NR2B, indicating that the IL3 region of D2R is involved in the interaction.
Next, to further explore the interaction sequence/site between D2R and NR2B, the IL3 region was segmented into: the specific positions of KVC (K211-V270) region of D2R, ES (E271-S321) region of D2R and PQ (P322-Q373) region of D2R on D2R are shown in FIG. 2.
According to the method of this example, pulldown analysis was performed using GST fusion protein (GST-D2R-KVC) containing the KVC region of D2R, GST fusion protein (GST-D2R-ES) containing the ES region of D2R, and GST fusion protein (GST-D2R-PQ) containing the PQ region of D2R, and the immunoblotting result is shown as B in FIG. 3.
From fig. 3B, it can be seen that the KVC region of D2R affinity precipitated NR2B, indicating that the KVC region of D2R is involved in the interaction.
Further, the KVC region is divided into: the KT (K211-T225) region of D2R, the KL (K226-L240) region of D2R, the KV (K241-V255) region of D2R and the IV (I256-V270) region of D2R, the specific positions of these regions on D2R are shown in FIG. 2.
According to the method of this example, a pulldown analysis was performed using a GST fusion protein containing the KT region of D2R (GST-D2R-KT), a GST fusion protein containing the KL region of D2R (GST-D2R-KL), a GST fusion protein containing the KV region of D2R (GST-D2R-KV) and a GST fusion protein containing the IV region of D2R (GST-D2R-IV), and the immunoblotting results are shown in C in FIG. 3.
Surprisingly, it is shown by C in fig. 3: the KL region of D2R (K226-L240) failed to precipitate NR2B, while the KT region of D2R (K211-T225, hereinafter referred to as KT polypeptide) affinity precipitated NR 2B.
Example 4 Polypeptides prevent Complex coupling in vivo
To verify the effect of the KT polypeptide of D2R on the D2R/NR2B complex in vivo, the C-terminus of the region (K211-T225) was fused to the N-terminus of the transduction domain of the HIV-1 type Tat protein (shown in SEQ ID NO:6, hereinafter referred to as TAT) to give a fusion protein that can penetrate the blood-brain barrier, named TAT-D2R-KT.
CPP mice and BS mice were treated with TAT-D2R-KT, respectively, and TAT-only treated mice were used as controls (both treatment were single intraperitoneal administration, 10ml/kg body weight, drug concentration 3 nmol/g). After 1 hour of treatment, the co-immunoprecipitation method in example 1 was followed for analysis, and the results are shown in FIG. 4.
As can be seen in FIG. 4, the KT polypeptide of the present invention competitively disrupted the interaction of D2R/NR2B in the hippocampal tissues of CPP mice and BS mice.
Example 5 polypeptide interference with morphine-induced CPP formation
To assess whether the polypeptides of the invention affect morphine addiction, CPP scores were measured using TAT-D2R-KT, TAT during the morphine-induced CPP formation phase.
The CPP equipment and test system are as follows:
the CPP apparatus used in the experiment had two equal volume chambers (15 cm. times.15 cm. times.37 cm) with a movable door in the middle. The mice were either able to shuttle freely between the chambers during testing or were confined to one side of the chamber during training. One chamber interior was black and the floor was striped, while the other chamber was white and the floor was checkered. All behavioral tests, including distance and time of activity, were recorded with a video camera and calculated using the SMART video tracking system (version 2.5; Panlab bio-retrieval technology, spain).
Experimental design as shown in figure 5, on day 1, mice were allowed to explore both chambers freely for 15min without any treatment (pre-test). On days 2, 4, 6, and 8, the control group (saline group) and the model group (morphine group) received saline (10mL/kg) and morphine (10mg/kg), respectively. TAT group (3nmol/g) and TAT-D2R-KT group (3nmol/g) were injected 1 hour before morphine injection (intraperitoneal administration, 10ml/kg body weight). After injection of the drug, all mice were trained in the white room for 40 min. On days 3, 5, 7, 9, all mice were given physiological saline and then immediately placed in the black room for 40 min; day 10, after 15min of free exploration of all mice) the test was ended and the CPP score (length of mouse activity) is shown in figure 6.
As can be seen in figure 6, TAT-D2R-KT treatment significantly reduced the CPP score compared to TAT-treated subjects, indicating that TAT-D2R-KT is able to interfere with morphine-induced CPP formation.
EXAMPLE 6 Effect of Polypeptides on morphine-induced CPP expression phase
To further evaluate the specific effect of the polypeptides of the invention on the morphine-induced CPP expression phase, tests were performed according to the experimental design in fig. 7, using the CPP device in example 5. Wherein the model group (morphine group) was injected with physiological saline on day 10, whereas the TAT-D2R-KT group (morphine + TAT-D2R-KT) treated CPP mice with TAT-D2R-KT (single dose) on day 10, CPP scores were measured after 1 hour, and the experimental results are shown as A in FIG. 8.
Treatment with TAT-D2R-KT at day 10 did not significantly reduce CPP scores, as shown by A in FIG. 8.
Nevertheless, by repeating the CPP test on day 11 we found: the CPP score in the TAT-D2R-KT group was significantly reduced at day 11 compared to day 10 (B in FIG. 8), indicating that TAT-D2R-KT might abrogate the associated memory at day 10 in morphine-addicted memory reproduction.
Example 7 Elimination of morphine-induced relapse by Polypeptides
To verify that the polypeptides of the invention were able to eliminate the associated memory in the recurrence of addictive memory and to test their long-term effect on morphine CPP relapse, we designed CPP mice natural withdrawal and relapse experiments as shown in fig. 9 and performed a number of tests with saline group as control.
Morphine successfully induced mouse CPP at day 10 according to the conditions shown in fig. 9, and the results are shown in fig. 10.
The mice were then housed for three weeks. On day 32, mice in the TAT group and TAT-D2R-KT group received TAT, TAT-D2R-KT (3nmol/g), respectively; after 1 hour, the mice were freely explored for 15min for test 1, and the results are shown in fig. 11.
As can be seen in FIG. 11, the CPP score was significantly reduced in mice of the TAT-D2R-KT group as compared to mice of the TAT group, indicating that the polypeptide of the present invention is able to eliminate the relapse caused by the environment.
To further explore the possible effect of polypeptides on addictive drug-induced relapse, after test 1, groups were kept in cages for three weeks and injected with morphine (5mg/kg) on day 54 in both TAT group and TAT-D2R-KT group, and all mice were immediately placed into the laboratory for free exploration for 15min for test 2, with the results shown in figure 12.
As can be seen in FIG. 12, CPP scores were again significantly reduced in mice of the TAT-D2R-KT group compared to mice of the TAT group.
To rule out the dose effect, groups were caged for two weeks after test 2 and mice in the TAT group and TAT-D2R-KT group were injected with higher doses of morphine (15mg/kg) on day 69, and then all mice were freely explored for 15 minutes for test 3, with the results shown in figure 13.
Surprisingly, higher doses of morphine still failed to reverse the effect of TAT-D2R-KT in significantly reducing CPP scores, suggesting that TAT-D2R-KT may abrogate low/high dose addictive substance-induced relapse.
To further explore the effect of TAT-D2R-KT on stress-induced relapse, groups were caged for one week after test 3 and tested 15 minutes after administration of yohimbine (α 2-adrenoceptor antagonist, 2mg/kg, from MCE, cat No. 18735) to mice in TAT group and TAT-D2R-KT group on day 77, as test 4, with the results shown in fig. 14.
As can be seen in FIG. 14, there was no significant difference in CPP score between mice in the TAT-D2R-KT group and the control group, indicating that the polypeptide of the present invention successfully blocks stress-induced relapse.
Example 8 verification of resistance to addiction-related behavior Using behavioral sensitization model
To further validate the effect of the polypeptides of the invention on morphine addiction related behavior, the above results were validated using a morphine-induced behavioral sensitization mouse (BS) model.
The experiment was performed according to the protocol shown in fig. 15, and all mice were habituated to the laboratory for 1 hour on day 1. Then, the mice were given physiological saline (control group) or 5mg/kg morphine (TAT group and TAT-D2R-KT group) and entered a laboratory (having a white surface and a flat bottom surface, and having dimensions of 50 cm. times.50 cm. times.35 cm)3) Locomotor activity was recorded immediately for 60min and the effect of the drug was assessed as a measure of the total distance traveled by the mice (denoted test 1), with the results shown in figure 16, a.
All mice were then reared for 1 week, and on day 9, mice were given TAT or TAT-D2R-KT (3nmol/g) 1 hour prior to morphine (1mg/kg) use, after which all mice were entered into the laboratory for testing to assess the effect of the drug (denoted test 2). The results shown in B in FIG. 16 show that 60-minute motor activity was significantly reduced in mice in the TAT-D2R-KT group.
After test 2, all mice were reared in cages for one week (weaning) and then re-entered the laboratory for testing (noted test 3). The results shown at C in figure 16 indicate that a significant reduction in locomotor activity could still be detected in the TAT-D2R-KT group after 1 week of withdrawal, further indicating that morphine-induced relapse could be abrogated by the polypeptide.
Example 9 design and preparation of polypeptide variants
Based on KT polypeptide (marked as Pep1), N-terminal deletion variant Pep2, C-terminal deletion variant Pep3, variant Pep4 of middle-section RRR mutated into GGG, and variant Pep5 of middle-section RKR mutated into GGG are respectively designed and synthesized, and are specifically shown in the following table 1:
TABLE 1
Name (R) Sequence of Type of mutation
Pep1(SEQ ID NO:1) KIYIVLRRRRKRVNT N.A.
Pep2(SEQ ID NO:3) YIVLRRRRKRVNT Truncation of the N-terminus
Pep3(SEQ ID NO:4) KIYIVLRRRRKRV Truncation of the C-terminus
Pep4(SEQ ID NO:7) KIYIVLGGGRKRVNT Middle RRR substituted
Pep5(SEQ ID NO:8) KIYIVLRRRGGGVNT Middle section RKR substituted
Then, the C ends of Pep 1-Pep 5 are respectively fused with the N end of TAT to obtain fusion peptide capable of penetrating blood brain barrier.
Example 10 demonstration of the Effect of different Polypeptides on morphine-induced mouse CPP
The mice were given morphine (1mg/kg) or physiological saline for 8 days, and the different polypeptides (3nmol/g) in example 9 were administered to the mice separately by intraperitoneal injection (10ml/kg) on day 10; morphine-induced CPP expression was recorded according to the CPP experimental method in example 5 on day 2 after administration, and the results are shown in fig. 17.
As can be seen from fig. 17, Pep1 and the truncated forms of Pep2 and Pep3 significantly reduced the distance of mobility of morphine CPP mice; whereas mid-segment substituted Pep4 and Pep5 failed to significantly reduce the distance of movement, suggesting that the key motif of the polypeptide is located in the middle of Pep 1.
Example 11 validation of the Effect of different polypeptides on ***e-induced mouse BS
To verify that the polypeptides of the invention are also effective against ***e-induced addiction, the following tests were performed:
mice were tested 30 minutes after ***e (15mg/kg) or saline administration for 5 days and then suspended for 5 days. The different polypeptides (3nmol/g) of example 9 were administered separately to mice by intraperitoneal injection (10ml/kg) 1 hour before ***e administration on the 6 th day of suspension, drug induction was performed and the results were tested as shown in fig. 18.
As can be seen from fig. 18, Pep1 and the truncated forms of Pep2 and Pep3 significantly inhibited the expression of sensitization behavior, in agreement with the expectation; whereas mid-substituted Pep4 and Pep5 failed to inhibit expression of sensitization behavior.
Example 12 validation of the Effect of different polypeptides on alcohol-induced mouse BS
To verify that the polypeptides of the invention are also effective against alcohol-induced addiction, the following tests were performed:
the training period was 10 days, and the mice were tested 15 minutes immediately after every other day of alcohol (2.2g/kg) or saline administration. On the 3 rd day after training, mice were administered the different polypeptides (3nmol/g) of example 9 separately by intraperitoneal injection (10ml/kg) 1 hour before alcohol administration, drug induction was performed and the test results were shown in fig. 19.
As can be seen from fig. 19, Pep1 and the truncated forms of Pep2 and Pep3 significantly inhibited the expression of sensitization behavior, in agreement with the expectation; whereas mid-substituted Pep4 and Pep5 failed to inhibit expression of sensitization behavior.
Sequence listing
<110> Shenzhen Yangyang Biotech Limited
<120> use of polypeptides for anti-addiction and relapse thereof, complexes and polypeptides
<130> 2113764-I-CP-CYSW
<160> 8
<170> PatentIn version 3.5
<210> 1
<211> 15
<212> PRT
<213> Artificial sequence
<400> 1
Lys Ile Tyr Ile Val Leu Arg Arg Arg Arg Lys Arg Val Asn Thr
1 5 10 15
<210> 2
<211> 11
<212> PRT
<213> Artificial sequence
<400> 2
Tyr Ile Val Leu Arg Arg Arg Arg Lys Arg Val
1 5 10
<210> 3
<211> 13
<212> PRT
<213> Artificial sequence
<400> 3
Tyr Ile Val Leu Arg Arg Arg Arg Lys Arg Val Asn Thr
1 5 10
<210> 4
<211> 13
<212> PRT
<213> Artificial sequence
<400> 4
Lys Ile Tyr Ile Val Leu Arg Arg Arg Arg Lys Arg Val
1 5 10
<210> 5
<211> 2482
<212> DNA
<213> Intelligent people
<400> 5
agagcctggc cacccagtgg ctccaccgcc ctgatggatc cactgaatct gtcctggtat 60
gatgatgatc tggagaggca gaactggagc cggcccttca acgggtcaga cgggaaggcg 120
gacagacccc actacaacta ctatgccaca ctgctcaccc tgctcatcgc tgtcatcgtc 180
ttcggcaacg tgctggtgtg catggctgtg tcccgcgaga aggcgctgca gaccaccacc 240
aactacctga tcgtcagcct cgcagtggcc gacctcctcg tcgccacact ggtcatgccc 300
tgggttgtct acctggaggt ggtaggtgag tggaaattca gcaggattca ctgtgacatc 360
ttcgtcactc tggacgtcat gatgtgcacg gcgagcatcc tgaacttgtg tgccatcagc 420
atcgacaggt acacagctgt ggccatgccc atgctgtaca atacgcgcta cagctccaag 480
cgccgggtca ccgtcatgat ctccatcgtc tgggtcctgt ccttcaccat ctcctgccca 540
ctcctcttcg gactcaataa cgcagaccag aacgagtgca tcattgccaa cccggccttc 600
gtggtctact cctccatcgt ctccttctac gtgcccttca ttgtcaccct gctggtctac 660
atcaagatct acattgtcct ccgcagacgc cgcaagcgag tcaacaccaa acgcagcagc 720
cgagctttca gggcccacct gagggctcca ctaaagggca actgtactca ccccgaggac 780
atgaaactct gcaccgttat catgaagtct aatgggagtt tcccagtgaa caggcggaga 840
gtggaggctg cccggcgagc ccaggagctg gagatggaga tgctctccag caccagccca 900
cccgagagga cccggtacag ccccatccca cccagccacc accagctgac tctccccgac 960
ccgtcccacc atggtctcca cagcactccc gacagccccg ccaaaccaga gaagaatggg 1020
catgccaaag accaccccaa gattgccaag atctttgaga tccagaccat gcccaatggc 1080
aaaacccgga cctccctcaa gaccatgagc cgtaggaagc tctcccagca gaaggagaag 1140
aaagccactc agatgctcgc cattgttctc ggcgtgttca tcatctgctg gctgcccttc 1200
ttcatcacac acatcctgaa catacactgt gactgcaaca tcccgcctgt cctgtacagc 1260
gccttcacgt ggctgggcta tgtcaacagc gccgtgaacc ccatcatcta caccaccttc 1320
aacattgagt tccgcaaggc cttcctgaag atcctccact gctgactctg ctgcctgccc 1380
gcacagcagc ctgcttccca cctccctgcc caggccggcc agcctcaccc ttgcgaaccg 1440
tgagcaggaa ggcctgggtg gatcggcctc ctcttcttag ccccggcagg ccctgcagtg 1500
ttcgcttggc tccatgctcc tcactgcccg cacaccctca ctctgccagg gcagtgctag 1560
tgagctgggc atggtaccag ccctggggct ggccccagct caggggcagc tcatagagtc 1620
ccccctccca cctccagtcc ccctatcctt ggcaccaaag atgcagccgc cttccttgac 1680
cttcctctgg ggctctaggg ttgctggagc ctgagtcagg gcccagaggc tgagttttct 1740
ctttgtgggg cttggcgtgg agcaggcggt ggggagagat ggacagttca caccctgcaa 1800
ggcccacagg aggcaagcaa gctctcttgc cgaggagcca ggcaacttca gtcctgggag 1860
acccatgtaa ataccagact gcaggttgga cccgagagat tcccaagcca aaaaccttag 1920
ctccctcccg caccccgatg tggacctcta ctttccaggc tagtccggac ccacctcacc 1980
ccgttacagc tccccaagtg gtttccacat gctctgagaa gaggagccct catcttgaag 2040
ggcccaggag ggtctatggg gagaggaact ccttggccta gcccaccctg ctgccttctg 2100
acggccctgc aatgtatccc ttctcacagc acatgctggc cagcctgggg cctggcaggg 2160
aggtcaggcc ctggaactct atctgggcct gggctaggga catcagaggt tctttgaggg 2220
actgcctctg ccacactctg acgcaaaacc actttccttt tctattcctt ctggcctttc 2280
ctctctcctg tttcccttcc cttccactgc ctctgcctta gaggagccca cggctaagag 2340
gctgctgaaa accatctggc ctggcctggc cctgccctga ggaaggaggg gaagctgcag 2400
cttgggagag cccctggggc ctagactctg taacatcact atccgatgca ccaaactaat 2460
aaaactttga cgagtcacct tc 2482
<210> 6
<211> 11
<212> PRT
<213> Artificial sequence
<400> 6
Tyr Gly Arg Lys Lys Arg Arg Gln Arg Arg Arg
1 5 10
<210> 7
<211> 15
<212> PRT
<213> Artificial sequence
<400> 7
Lys Ile Tyr Ile Val Leu Gly Gly Gly Arg Lys Arg Val Asn Thr
1 5 10 15
<210> 8
<211> 15
<212> PRT
<213> Artificial sequence
<400> 8
Lys Ile Tyr Ile Val Leu Arg Arg Arg Gly Gly Gly Val Asn Thr
1 5 10 15

Claims (10)

1. Use of a polypeptide for the preparation of a medicament for the treatment and/or prevention of substance addiction, wherein the polypeptide consists of at least 11 consecutive amino acid residues of the sequence as shown in SEQ ID No. 1 and comprises the sequence as shown in SEQ ID No. 2.
2. Use of a polypeptide for the manufacture of a medicament for the treatment and/or prevention of relapse of substance addiction, wherein the polypeptide consists of at least 11 consecutive amino acid residues of the sequence as shown in SEQ ID No. 1 and comprises the sequence as shown in SEQ ID No. 2.
3. The use of claim 2, wherein relapse is caused by environmental factors, addictive substance factors, or stress factors.
4. Use according to any one of claims 1 to 3, wherein the amino acid sequence of the polypeptide is selected from the group consisting of the sequences shown as SEQ ID NO 1, SEQ ID NO 3 and SEQ ID NO 4.
5. Use according to any one of claims 1 to 4, wherein the substance inducing substance addiction is selected from one or more of the group consisting of: morphine, nicotine, alcohol, ***e, codeine, dihydrocodeine, hydromorphone, oxycodone, methadone, morphine, fentanyl, and meperidine.
6. Use of a nucleic acid molecule for the manufacture of a medicament for the treatment and/or prevention of substance addiction or relapse of substance addiction, wherein the nucleic acid molecule encodes a polypeptide as defined in any one of claims 1 to 5.
7. Use of an expression vector in the manufacture of a medicament for the treatment and/or prevention of substance addiction or relapse of substance addiction, wherein the expression vector comprises the nucleic acid molecule of claim 6.
8. Use of a host cell in the manufacture of a medicament for the treatment and/or prevention of substance addiction or relapse of substance addiction, wherein the host cell comprises the nucleic acid molecule of claim 6 or the expression vector of claim 7.
9. A complex comprising a polypeptide as defined in any one of claims 1 to 5 and a carrier for permeabilizing the blood-brain barrier attached thereto, optionally the carrier for permeabilizing the blood-brain barrier is selected from the group consisting of: HIV-1Tat protein, insulin, cationized albumin, monoclonal antibody against mouse transferrin receptor, murine monoclonal antibody against human insulin receptor, Pentratin, transduction domain of Tat protein, Pep-1 peptide, S413-PV, Magainin 2 and Buforin 2.
10. A polypeptide consisting of at least 11 and at most 14 consecutive amino acid residues of the sequence shown as SEQ ID NO. 1 and comprising the sequence shown as SEQ ID NO. 2.
CN202111321033.2A 2021-11-09 2021-11-09 Anti-addiction and anti-relapse application of polypeptide, complex and polypeptide Pending CN113813364A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202111321033.2A CN113813364A (en) 2021-11-09 2021-11-09 Anti-addiction and anti-relapse application of polypeptide, complex and polypeptide
PCT/CN2022/117272 WO2023082805A1 (en) 2021-11-09 2022-09-06 Use of polypeptide in resistance to addiction and relapse thereof, and complex and polypeptide

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111321033.2A CN113813364A (en) 2021-11-09 2021-11-09 Anti-addiction and anti-relapse application of polypeptide, complex and polypeptide

Publications (1)

Publication Number Publication Date
CN113813364A true CN113813364A (en) 2021-12-21

Family

ID=78919487

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111321033.2A Pending CN113813364A (en) 2021-11-09 2021-11-09 Anti-addiction and anti-relapse application of polypeptide, complex and polypeptide

Country Status (2)

Country Link
CN (1) CN113813364A (en)
WO (1) WO2023082805A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023082805A1 (en) * 2021-11-09 2023-05-19 深圳辰扬生物科技有限公司 Use of polypeptide in resistance to addiction and relapse thereof, and complex and polypeptide
CN116514924A (en) * 2023-06-02 2023-08-01 徐州医科大学 Polypeptide for relieving mouse morphine addiction and application thereof
WO2023202663A1 (en) * 2022-04-21 2023-10-26 深圳辰扬生物科技有限公司 Polypeptide and use thereof

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005055942A2 (en) * 2003-12-05 2005-06-23 University Of Massachusetts Modulation of brain pathways and function
US20080213271A1 (en) * 2005-05-05 2008-09-04 Center For Addiction And Mental Health Compositions and Methods For Modulating Dopamine Nerutransmission
US20120093822A1 (en) * 2009-04-09 2012-04-19 Centre Forr Addiction And Mental Health Dopamine d2 receptor-disc1 interaction, compositions and methods for modulating same
US20150297742A1 (en) * 2012-12-05 2015-10-22 Ruprecht-Karls-Universitat Conjugates of Proteins and Multivalent Cell-Penetrating Peptides and Their Uses
US20170027972A1 (en) * 2015-06-09 2017-02-02 The Royal Institution For The Advancement Of Learning / Mcgill University Methods and Compositions for Treatment of Drug Addiction
CN109106942A (en) * 2018-09-18 2019-01-01 北京大学深圳研究生院 It is a kind of to pass through the application of the polypeptide of blood-brain barrier in medicine preparation
CN111606976A (en) * 2020-05-26 2020-09-01 中国人民解放军军事科学院军事医学研究院 Small peptide and application thereof in inhibiting opiate addiction and tolerance

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109553687B (en) * 2017-09-27 2021-07-06 北京大学 Fluorescent probe constructed based on G protein coupled receptor
CN113813364A (en) * 2021-11-09 2021-12-21 深圳辰扬生物科技有限公司 Anti-addiction and anti-relapse application of polypeptide, complex and polypeptide

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005055942A2 (en) * 2003-12-05 2005-06-23 University Of Massachusetts Modulation of brain pathways and function
US20080213271A1 (en) * 2005-05-05 2008-09-04 Center For Addiction And Mental Health Compositions and Methods For Modulating Dopamine Nerutransmission
US20120093822A1 (en) * 2009-04-09 2012-04-19 Centre Forr Addiction And Mental Health Dopamine d2 receptor-disc1 interaction, compositions and methods for modulating same
US20150297742A1 (en) * 2012-12-05 2015-10-22 Ruprecht-Karls-Universitat Conjugates of Proteins and Multivalent Cell-Penetrating Peptides and Their Uses
US20170027972A1 (en) * 2015-06-09 2017-02-02 The Royal Institution For The Advancement Of Learning / Mcgill University Methods and Compositions for Treatment of Drug Addiction
CN109106942A (en) * 2018-09-18 2019-01-01 北京大学深圳研究生院 It is a kind of to pass through the application of the polypeptide of blood-brain barrier in medicine preparation
CN111606976A (en) * 2020-05-26 2020-09-01 中国人民解放军军事科学院军事医学研究院 Small peptide and application thereof in inhibiting opiate addiction and tolerance

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ANDRY ANDRIANARIVELO等: "Disrupting D1-NMDA or D2-NMDA receptor heteromerization prevents ***e’s rewarding effects but preserves natural reward processing", 《SCIENCE ADVANCES》, vol. 43, no. 7, pages 1 - 17 *
ANDRY ANDRIANARIVELO等: "Disrupting D2-NMDA receptor heteromerization blocks the rewarding effects of ***e but preserves natural reward processing", 《HTTPS://DOI.ORG/10.1101/2021.01.25.428078》, pages 1 - 36 *
XIANYU LIU等: "Modulation of D2R-NR2B Interactions in Response to Cocaine", 《NEURON》, vol. 52, pages 897 *
李凌江: "行为医学", 31 August 2016, 长沙:湖南科学技术出版社, pages: 104 - 108 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023082805A1 (en) * 2021-11-09 2023-05-19 深圳辰扬生物科技有限公司 Use of polypeptide in resistance to addiction and relapse thereof, and complex and polypeptide
WO2023202663A1 (en) * 2022-04-21 2023-10-26 深圳辰扬生物科技有限公司 Polypeptide and use thereof
CN116514924A (en) * 2023-06-02 2023-08-01 徐州医科大学 Polypeptide for relieving mouse morphine addiction and application thereof
CN116514924B (en) * 2023-06-02 2023-11-24 徐州医科大学 Polypeptide for relieving mouse morphine addiction and application thereof

Also Published As

Publication number Publication date
WO2023082805A1 (en) 2023-05-19

Similar Documents

Publication Publication Date Title
CN113813364A (en) Anti-addiction and anti-relapse application of polypeptide, complex and polypeptide
US8722851B2 (en) Analgesia with minimal tolerance and dependence by a mu opioid receptor agonist that also binds filamin A
AU739263B2 (en) Antagonists of intestinotrophic GLP-2 peptides
JP2001520634A (en) Opioid antagonists and methods of using them
JP2020500897A (en) Modulators of complement activity
JP2002509939A (en) Novel peptides for the treatment, prevention, diagnosis and monitoring of autoimmune diseases
AU713546B2 (en) Peptide specificity of anti-myelin basic protein and the administration of myelin basic protein peptides to multiple sclerosis patients
EP3524257A1 (en) Use of excitatory nerve injury-related polypeptide in preventing, alleviating or treating pain
CN113855784A (en) Anti-depression and anti-anxiety application of polypeptide and complex
CA2423073A1 (en) Recombinant antibody fragments as autoantibody antagonists
WO2023202663A1 (en) Polypeptide and use thereof
AU2003288434B2 (en) Peptides, antibodies thereto, and their use in the treatment of central nervous system damage
CN113474363A (en) GDF15 analogs and methods for reducing body weight and/or reducing food intake
DK2828285T3 (en) HB-EGF INHIBITOR DERIVED FROM DIFTERY TOXIN R DOMAIN FOR THE TREATMENT OF DISEASES ASSOCIATED WITH THE ACTIVATION OF HB-EGF / EGFR SIGNAL ROAD
DE69928523T2 (en) VEROTOXIN B SUB-UNIT FOR IMMUNIZATION
CN111848805A (en) Bispecific antibodies with dual Her2 sites for tumor immunotherapy
CN111225681A (en) Method of producing a composite material
US20230416326A1 (en) Modified interleukin-2 (il-2) molecule and use thereof
Ricci et al. Helicobacter pylori vacuolating toxin
EP1909816B1 (en) Cnf1 bacterial toxin protein from e. coli as analgesic
Itzhak et al. A novel delta opioid receptor specific peptide reduces craving in an animal model of ***e seeking
JP2001504451A (en) Peptides as diagnostic and therapeutic agents for autoimmune diseases
WO2020251936A1 (en) COMPOSITIONS AND METHODS FOR INHIBITING THE BINDING OF PLASMA IgG AUTOANTIBODIES TO SEROTONIN 2A RECEPTOR
Grinnell Truncated mu opioid receptor splice variants as targets for powerful pain relief with reduced side effects
CN106344931A (en) Coupling conjugate of micromolecular amyloid antibody polypeptide and estrogen

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination